CN116143843A - Metal complex with FTO inhibition activity and preparation method and application thereof - Google Patents

Metal complex with FTO inhibition activity and preparation method and application thereof Download PDF

Info

Publication number
CN116143843A
CN116143843A CN202310142825.6A CN202310142825A CN116143843A CN 116143843 A CN116143843 A CN 116143843A CN 202310142825 A CN202310142825 A CN 202310142825A CN 116143843 A CN116143843 A CN 116143843A
Authority
CN
China
Prior art keywords
fto
metal complex
dimer
metal
inhibitory activity
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202310142825.6A
Other languages
Chinese (zh)
Inventor
刘红科
黄元镭
薛旭玲
吕梦迪
郭冰莲
胡国静
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nanjing Normal University
Original Assignee
Nanjing Normal University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanjing Normal University filed Critical Nanjing Normal University
Priority to CN202310142825.6A priority Critical patent/CN116143843A/en
Publication of CN116143843A publication Critical patent/CN116143843A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F15/00Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table
    • C07F15/0006Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table compounds of the platinum group
    • C07F15/0046Ruthenium compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F15/00Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table
    • C07F15/0006Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table compounds of the platinum group
    • C07F15/002Osmium compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F15/00Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table
    • C07F15/0006Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table compounds of the platinum group
    • C07F15/0033Iridium compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F15/00Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table
    • C07F15/0006Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic Table compounds of the platinum group
    • C07F15/0073Rhodium compounds

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)

Abstract

The invention discloses a metal complex with FTO inhibition activity, and also discloses a preparation method and application of the metal complex with FTO inhibition activity. The metal complex is obtained by reacting an alkynyl modified FTO inhibitor with a metal complex precursor containing an azide group through a CuAAC reaction, has good anticancer activity and FTO inhibition activity, can overcome the problem of poor target selectivity of the FTO inhibitor alone on one hand, and can overcome the problem of large toxic and side effects when the pure metal complex (precursor) is used as an antitumor drug or an antitumor drug component on the other hand.

Description

Metal complex with FTO inhibition activity and preparation method and application thereof
Technical Field
The invention relates to a metal complex with FTO inhibition activity, and also relates to a preparation method and application of the metal complex.
Technical Field
Cancer is one of the most serious diseases that threaten human life at present. Most of the platinum and other drugs (such as cisplatin, carboplatin, oxaliplatin and the like) used in clinic at present have the defects of large toxic and side effects, poor selectivity, drug resistance and the like. With the deep research of the action mechanism of anticancer drugs in tumor cells, in recent years, metal antitumor drugs with a wide application prospect have appeared. At present, metal complexes such as aryl ruthenium, iridium, rhodium, osmium and the like are considered as the most promising metal anticancer drugs because of the advantages of high activity, selectivity, multiple targets, low drug resistance and the like. At present, many specialists at home and abroad, such as professor Peter Sadler at university of wawei, united kingdom, professor Paul Dyson at the university of Vienna in Switzerland, professor Bernhard Keppler, et al, have been studied intensively in this regard.
In 2007, UK scientists found the FTO gene (Fat mass and obesity-associated protein) related to obesity in Single Nucleotide Polymorphism (SNP) study related to type II diabetes mellitus, and the FTO gene coding protein is a member of AlkB family and depends on ferrous Fe 2+ And 2-oxoglutarate (2 OG) has the function of catalyzing the demethylation of nucleotides. Related studies have shown that FTO plays an oncogene role in the development of acute myelogenous leukemia and clearly reveals an m on FTO-mediated mRNA 6 The A modification is involved in the development of tumors. Researchers in Shanghai pharmaceutical institute 2012 screen out 3 compounds such as rhein which can inhibit the activity of FTO, wherein rhein has the best inhibition effect and is more suitable for serving as candidate drug molecules. In 2014, american scientists unexpectedly found that the obtained chemical can not effectively inhibit target protein PHD2, but can effectively inhibit FTO of 2OG oxygenase family, and IC 50 The values were close to the broad-spectrum 2OG enzyme inhibitor NOG, and stronger than rhein reported previously. However, most of the FTO inhibitors reported so far are unsuitable for clinical use due to poor target selectivity or poor pharmacokinetics.
Disclosure of Invention
The invention aims to: the invention aims to provide a metal complex with FTO inhibition activity, which is obtained by reacting an alkynyl modified FTO inhibitor with a metal complex precursor containing an azide group through a CuAAC reaction, and has good anticancer activity and FTO inhibition activity, so that on one hand, the problem of poor target selectivity of the single FTO inhibitor can be overcome, and on the other hand, the problem of great toxic and side effects when the single metal complex (precursor) is used as an antitumor drug or an antitumor drug component can be overcome.
The technical scheme is as follows: the structural general formula of the metal complex with FTO inhibition activity is shown as follows:
Figure BDA0004088110750000021
wherein M is Ru, ir, rh or Os;
Figure BDA0004088110750000022
is->
Figure BDA0004088110750000023
L is
Figure BDA0004088110750000024
The preparation method of the metal complex with the FTO inhibition activity comprises the following steps:
(1) Preparation of azide group-containing metal complex precursors: under inert atmosphere, dissolving a metal ruthenium dimer, a metal iridium dimer, a metal rhodium dimer or a metal osmium dimer in methanol, carrying out coordination reaction with a chelating ligand, removing a solvent by reduced pressure rotary evaporation, adding a methanol solution of saturated anion salt, freezing at low temperature, centrifuging after freezing, collecting solids, and purifying a crude product by column chromatography to obtain a metal complex precursor;
(2) Preparation of alkynyl-modified organic active molecule ligands: under inert atmosphere, placing organic active molecules, alkynyl compounds and catalysts into an organic solvent for reaction, removing the solvent by rotary evaporation after the reaction is finished, and purifying a crude product by column chromatography to obtain an organic active molecule ligand;
(3) Under inert atmosphere, the metal complex precursor, the organic active molecular ligand and the catalyst are subjected to CuAAC reaction in an organic solvent, after the reaction is finished, the solvent is removed by rotary evaporation, and the crude product is purified by a chromatographic column to obtain the metal complex.
In the step (1), the metal ruthenium dimer is p-cymene ruthenium (II) dichloride dimer (available from Shanghai Michelin Biochemical technologies Co., ltd., CAS: 52462-29-0), the metal iridium dimer is dichloro (pentamethylcyclopentadienyl) iridium (III) dimer (available from Shanghai Michelin Biochemical technologies Co., ltd., CAS: 12354-84-6), the metal rhodium dimer is dichloro (pentamethylcyclopentadienyl) rhodium (III) dimer (available from Shanghai Michelin Biochemical technologies Co., ltd., CAS: 12354-85-7), and the metal osmium dimer is p-cymene osmium (II) dichloride dimer (available from aablocks Co., CAS: 78615-08-4).
Wherein, in the step (1), the molar ratio of the dimer of the metallic ruthenium to the chelating ligand is 8-9: 1, a step of; the molar ratio of the dimer of the metallic iridium to the chelating ligand is 13-14: 1, a step of; the molar ratio of the dimer of the metal rhodium to the chelating ligand is 12-13: 1, a step of; the molar ratio of the dimer of the metal osmium to the chelating ligand is 12-13: 1.
wherein in the step (1), the chelating ligand is 4-azidomethyl-4 '-methyl-2, 2' -bipyridine, and the 4-azidomethyl-4 '-methyl-2, 2' -bipyridine is prepared by a method reported in the document Artificial photosynthesis dendrimers integrating light-harvesting, electron delivery and hydrogen production.
Wherein, in the step (1), the freezing temperature is between-20 and 0 ℃ and the freezing time is between 8 and 14 hours.
Wherein in step (2), the alkynyl compound is bromopropyne, propargylamine or pentynoic acid; the catalyst is potassium carbonate or N-hydroxybenzotriazole and N, N-diisopropylethylamine according to the mol ratio of 82-83: 135, and a combination thereof.
Wherein in the step (2), the molar ratio of the organic active molecule to the alkynyl compound is 0.1-4: 1.
wherein in the step (3), the catalyst is cupric sulfate pentahydrate and sodium ascorbate according to the mole ratio of 3-3.5: 4.
Wherein in the step (3), the reaction mole ratio of the metal complex precursor and the organic active molecular ligand is 3-25.5: 1.
the metal complex with the FTO inhibition activity is applied to the preparation of antitumor drugs, antitumor drug components, FTO inhibition drugs and FTO inhibition drug components.
The reaction mechanism of the metal complex synthesis process is as follows:
Figure BDA0004088110750000031
wherein:
Figure BDA0004088110750000032
the metal complex is a metal complex precursor, and the structural formula of the metal complex precursor is as follows; />
Figure BDA0004088110750000033
X is an azide group in the metal complex precursor;
wherein: m is Ru, ir, rh or Os;
wherein:
Figure BDA0004088110750000041
is p-cymene or pentamethyl cyclopentadiene;
wherein:
Figure BDA0004088110750000042
is an organic active molecular ligand, and has the structural formula: />
Figure BDA0004088110750000043
Figure BDA0004088110750000044
Y is an alkyne group in the organic reactive molecular ligand.
The aryl/metallocene metal complex is obtained by taking a molecule with anti-FTO activity as an alkyne end and taking aryl/metallocene metal as an azide end through CuAAC reaction, has good fat solubility, can improve active oxygen generated by cancer cells, induces the generation of mitochondrial membrane potential and blocks cell cycle.
The beneficial effects are that: according to the invention, the FTO inhibitor with anti-tumor, antibacterial and anti-inflammatory effects is subjected to specific group modification and coordinated with cyclopentadienyl and aryl metal, so that a complex with good obesity gene FTO activity, anti-cancer activity, antibacterial activity and anti-inflammatory activity is obtained, and therefore, the metal complex has the anti-cancer, antibacterial, anti-inflammatory and FTO demethylase inhibiting activities with targeting effects.
Drawings
FIG. 1 is a graph showing the cell localization of the complex FTO-2-Ru/Ir/Rh in cells;
FIG. 2 is a diagram of apoptosis induced by a complex FTO-2-Ru/Ir/Rh, an organic active molecule ligand and an organic active molecule;
FIG. 3 is a confocal fluorescence imaging and flow cytometry of the complex FTO-2-Ru/Ir/Rh with organic active molecule ligands and organic active molecules to promote intracellular reactive oxygen species generation;
FIG. 4 is a flow cytometry and confocal fluorescence imaging of the complexes FTO-2-Ru/Ir/Rh with organic active molecule ligands and organic active molecules inducing a decrease in mitochondrial membrane potential;
FIG. 5 is a graph showing the effect of the complex FTO-2-Ru/Ir/Rh on cell cycle arrest of organic active molecule ligands and organic active molecules.
FIG. 6 is a schematic representation of the FTO activity inhibition by the complex FTO-2-Ru/Ir/Rh, organic active molecule ligands and organic active molecules.
Detailed Description
The technical scheme of the invention is further described below with reference to specific embodiments.
Example 1
The preparation method of the metal complex FTO-2-Ru comprises the following steps:
Figure BDA0004088110750000051
step 1, 6- (5-methylpyrazin-2-yl) naphthalen-2-ol (FTO-2) (0.13 g,0.5 mmol), bromopropyne (0.53 g,4.5 mmol) and potassium carbonate (2.05 g,15.2 mmol) were dissolved in DMF (N, N-dimethylformamide), stirred at 80℃for 12h, water was added, extracted with ethyl acetate, dried over anhydrous sodium sulphate, the solvent was removed by rotary evaporation to give the crude product which was isolated and purified by column chromatography to give the organic compound FTO-2-alkyne in 85% yield. 1 H NMR(400MHz,DMSO-d 6 )δ9.06(s,2H),8.25(d,J=1.9Hz,1H),7.93(dd,J=8.8,6.7Hz,2H),7.86(dd,J=8.6,1.9Hz,1H),7.45(d,J=2.6Hz,1H),7.26(dd,J=9.0,2.6Hz,1H),4.95(d,J=2.4Hz,2H),3.99(s,3H),3.64(t,J=2.4Hz,1H).
The FTO-2 was prepared by the synthetic method reported in document Broad spectrum anti-cancer compounds for treatment of cancer.
Step 2, at ambient temperature, the bridged ligand 4-azidomethyl-4 '-methyl-2, 2' -bipyridine (0.9 g,4 mmol) was reacted with p-cymene ruthenium (II) dichloride dimer (1.96 g,3.2 mmol) in methanol solution for 48h, the solvent was removed by rotary evaporation, and saturated (NH) was added 4 )PF 6 Freezing at-20deg.C in refrigerator overnight, centrifuging to collect solid, purifying the crude product by column chromatography to obtain pure product with yield of 85% and designated Ru-N 31 H NMR(400MHz,DMSO-d 6 )δ9.50(d,J=5.8Hz,1H),9.36(d,J=5.8Hz,1H),8.60–8.57(m,2H),7.74(dd,J=6.0,1.7Hz,1H),7.67–7.63(m,1H),6.22–6.19(m,2H),5.98–5.94(m,2H),4.88(s,2H),2.58(s,3H),2.57–2.52(m,1H),2.18(s,3H),0.94(dd,J=7.0,2.0Hz,6H).
The bridged ligand 4-azidomethyl-4 '-methyl-2, 2' -bipyridine is prepared by adopting a synthesis method reported in the document Artificial photosynthesis dendrimers integrating light-harvesting, electron delivery and hydrogen production, and is specifically as follows:
4,4 '-dimethyl-2, 2' -bipyridine (8.0 g,4.3 mmol) and 1, 4-dioxane (50-250 mL) were added to prepare a suspension, and SeO was added 2 (8.0 g,7.1 mmol) was heated under reflux for 48h, filtered to give a filtrate, and the solvent was removed under reduced pressure to give a solid; dissolving the solid substances in chloroform, filtering, removing the filtrate under reduced pressure to obtain solid substances, and repeating the steps for more than three times to finally obtain a crude product; sodium borohydride (2.8 g) was dissolved in sodium hydroxide solution (0.2 mol,20 mL), added dropwise to a methanol suspension (50 mL) in which the crude product was dissolved, stirred for 1h under cooling, methanol was removed under reduced pressure, and 80mL of saturated Na was added 2 CO 3 Diluting the solution, extracting the dried organic phase, evaporating the solvent, and purifying by chromatography to obtain 4-hydroxymethyl-4 '-methyl-2, 2' -bipyridine; 4-hydroxymethyl-4 '-methyl-2, 2' -bipyridine (3.2 g,5.3 mmol) was dissolved in HBr (48%, 40 mL), concentrated sulfuric acid (10 mL) was added, heated and refluxed overnight, cooled, then the pH was adjusted to 8, extracted with chloroform until the organic layer was colorless, and the organic layer was dried to remove chloroform, to give 4-bromomethyl-4 '-methyl-2, 2' -bipyridine; 4-bromomethyl-4 '-methyl-2, 2' -bipyridine (2.8 g,10.7 mmol) and NaN 3 (1.2 g,18.5 mmol) was dissolved in aqueous N, N-dimethylformamide (55 mL, v/v=10:1) and stirred at 70℃for 12h, the solvent was removed to give the crude product, which was used with CH 2 Cl 2 The organic layer obtained by extraction was washed with water, dried, and then the solvent was removed, and purified by chromatography to obtain 4-azidomethyl-4 '-methyl-2, 2' -bipyridine. 1 H NMR(400MHz,DMSO-d 6 )δ8.68(dd,J=4.9,0.8Hz,1H),8.55(dd,J=5.0,0.8Hz,1H),8.38(dd,J=1.7,0.9Hz,1H),8.26(dt,J=1.7,0.9Hz,1H),7.44–7.41(m,1H),7.31(ddd,J=5.0,1.8,0.8Hz,1H),4.70(s,2H),2.42(d,J=0.7Hz,3H).
Step 3, FTO-2-alkyne (0.52 g,1.8 mmol) obtained in step 1 and Ru-N were subjected to argon atmosphere 3 (1.49 g,3 mmol), copper sulfate pentahydrate (0.34 g,1.50 mmol) and sodium ascorbate (0.40 g,2.0 mmol) were dissolved in DMF and stirred for 10h, after the reaction was completed, the solvent was removed by rotary evaporation, and the orange-yellow product FTO-2-Ru was obtained by column chromatography separation in 83.3% yield. 1 H NMR(400MHz,DMSO-d 6 )δ9.50(d,J=5.9Hz,1H),9.37(d,J=5.8Hz,1H),9.06(s,2H),8.64(s,1H),8.54(s,1H),8.48(s,1H),8.25(s,1H),7.97–7.90(m,2H),7.89–7.83(m,1H),7.66(d,J=5.9Hz,1H),7.58(d,J=2.5Hz,1H),7.49(d,J=5.9Hz,1H),7.26(dd,J=8.9,2.5Hz,1H),6.19(dd,J=14.8,6.1Hz,2H),6.00–5.91(m,4H),5.35(s,2H),3.99(s,3H),2.58(s,3H),2.55(s,1H),2.16(s,3H),0.94(t,J=7.0Hz,6H).
Example 2
The preparation method of the metal complex FTO-2-Ir comprises the following steps:
Figure BDA0004088110750000061
step 1, at ambient temperature, the bridged ligand 4-azidomethyl-4 '-methyl-2, 2' -bipyridine (0.9 g,4 mmol) was reacted with dichloro (pentamethylcyclopentadienyl) iridium (III) dimer (2.54 g,3.2 mmol) in methanol solution for 10h, the solvent was removed by rotary evaporation, and saturated (NH) was added 4 )PF 6 Freezing at-20deg.C in refrigerator overnight, centrifuging to collect solid, purifying the crude product by column chromatography to obtain pure product with yield of 87.6% and designated Ir-N 31 H NMR(400MHz,DMSO-d 6 )δ8.95(d,J=5.8Hz,1H),8.81(d,J=5.8Hz,1H),8.72(t,J=2.3Hz,2H),7.79(dd,J=6.0,1.7Hz,1H),7.70(dd,J=6.0,1.7Hz,1H),4.94(s,2H),2.64(s,3H),1.65(s,15H).
Step 2 FTO-2-Alkyne (0.52 g,1.8 mmol) of step 1 of example 1, ir-N under argon atmosphere 3 (2.17 g,3.0 mmol), copper sulfate pentahydrate (0.34 g,1.50 mmol) and sodium ascorbate (0.40 g,2.0 mmol) were dissolved in DMF and stirred for 10h, after the reaction was completed, the solvent was removed by rotary evaporation, and the orange-yellow product FTO-2-Ir was obtained by column chromatography separation in 85.3% yield. 1 H NMR(400MHz,DMSO-d 6 )δ9.06(s,2H),8.95(d,J=5.9Hz,1H),8.84–8.78(m,2H),8.62(s,1H),8.55(s,1H),8.25(d,J=1.8Hz,1H),7.96–7.91(m,2H),7.86(dd,J=8.6,1.9Hz,1H),7.71(d,J=5.8Hz,1H),7.59(d,J=2.6Hz,1H),7.49(dd,J=5.9,1.8Hz,1H),7.27(dd,J=8.9,2.5Hz,1H),6.00(s,2H),5.36(s,2H),3.99(s,3H),2.64(s,3H),1.64(s,15H).
Example 3
The preparation method of the metal complex FTO-2-Rh comprises the following steps:
Figure BDA0004088110750000071
step 1, at ambient temperature, the bridging ligand 4-azidomethyl-4 '-methyl-2, 2' -bipyridine (0.9 g,4 mmol) was reacted with dichloro (pentamethylcyclopentadienyl) rhodium (III) dimer (2.26 g,3.2 mmol) in methanol solution for 10h, the solvent was removed by rotary evaporation, and saturated (NH) was added 4 )PF 6 Freezing at-20deg.C in refrigerator overnight, centrifuging to collect solid, purifying the crude product by column chromatography to obtain pure product with yield of 78.5%, designated Rh-N 31 H NMR(400MHz,DMSO-d 6 )δ8.95(d,J=5.7Hz,1H),8.81(d,J=5.7Hz,1H),8.62(t,J=1.9Hz,2H),7.82(dd,J=5.8,1.7Hz,1H),7.75–7.69(m,1H),4.89(s,2H),2.59(s,3H),1.66(s,15H).
Step 2 FTO-2-Alkyne (0.52 g,1.8 mmol) of step 1 of example 1, rh-N under argon atmosphere 3 (1.91 g,3 mmol), copper sulfate pentahydrate (0.34 g,1.50 mmol) and sodium ascorbate (0.40 g,2.0 mmol) were dissolved in DMF and stirred for 10h, after the reaction was completed, the solvent was removed by rotary evaporation, and the column chromatography separation gave the orange-yellow product FTO-2-Rh with a yield of 86.5%. 1 H NMR(400MHz,DMSO-d 6 )δ9.06(s,2H),8.95(d,J=5.8Hz,1H),8.82(d,J=5.7Hz,1H),8.72(s,1H),8.54(d,J=4.8Hz,2H),8.25(s,1H),7.98–7.91(m,2H),7.88–7.84(m,1H),7.73(d,J=5.8Hz,1H),7.59(d,J=2.5Hz,1H),7.52(d,J=5.8Hz,1H),7.27(dd,J=8.9,2.4Hz,1H),5.96(s,2H),5.36(s,2H),3.99(d,J=1.1Hz,3H),2.59(s,3H),1.65(s,15H).
Example 4
The preparation method of the metal complex FTO-2-Os comprises the following steps:
Figure BDA0004088110750000081
step 1, at ambient temperature, the bridged ligand 4-azidomethyl-4 '-methyl-2, 2' -bipyridine (0.9 g,.4 mmol) was reacted with osmium (II) dimer (2.25 g,3.2 mmol) in methanol solution for 10h, the solvent was removed by rotary evaporation, and saturated (NH) was added 4 )PF 6 Freezing at-20deg.C in refrigerator overnight, centrifuging to collect solid, purifying the crude product by column chromatography to obtain pure product with 86.3% yield, designated as Os-N 31 H NMR(400MHz,CD 3 OD)δ9.21(d,J=5.9Hz,1H),9.02(d,J=5.8Hz,1H),8.33(d,J=19.1Hz,2H),7.52(ddd,J=44.9,5.9,1.4Hz,2H),6.31-6.15(m,2H),5.74-5.59(m,2H),4.81(s,2H),2.58(m,4H),2.23(s,3H),1.11(dd,J=6.9,1.0Hz,6H).
Step 2 under argon atmosphere, FTO-2-Alkyne (0.52 g,1.8 mmol) of step 1 of example 1, os-N 3 (1.76 g,3 mmol), copper sulfate pentahydrate (0.34 g,1.50 mmol) and sodium ascorbate (0.40 g,2.0 mmol) were dissolved in DMF and stirred for 10h, after the reaction was completed, the solvent was removed by rotary evaporation, and the orange-yellow product FTO-2-Os was obtained by column chromatography separation in 84.2% yield. 1 H NMR(400MHz,DMSO-d 6 )δ9.52(d,J=5.9Hz,1H),9.38(d,J=5.8Hz,1H),9.07(s,2H),8.63(s,1H),8.52(s,1H),8.46(s,1H),8.26(s,1H),7.97–7.91(m,2H),7.89–7.85(m,1H),7.63(d,J=5.9Hz,1H),7.57(d,J=2.5Hz,1H),7.45(d,J=5.9Hz,1H),7.26(dd,J=8.9,2.5Hz,1H),6.18(dd,J=14.8,6.1Hz,2H),6.02–5.90(m,4H),5.32(s,2H),3.95(s,3H),2.59(s,3H),2.56(s,1H),2.26(s,3H),1.14(t,J=7.0Hz,6H).
Example 5
The preparation method of the metal complex FTO-4-Ru comprises the following steps:
Figure BDA0004088110750000091
step 1, pentynoic acid (29.4 mg,0.3 mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (57.5 mg,0.3 mmol) and triethylamine (50. Mu.L) were dissolved in DMF under argon atmosphere and ice-bathed in stripsN-hydroxybenzotriazole (32.4 mg,0.24 mmol) was dissolved in DMF and added dropwise to the above mixture, reacted under ice bath for 15min, then 6- (5-methoxypyridin-3-yl) benzothiazol-2-amine (FTO-4) (129.2 mg,0.5 mmol) was added dropwise to the mixed solution, the reaction mass was allowed to warm to room temperature, after stirring overnight, the organic solvent was removed, and purified by column chromatography to give FTO-4-alkyne as a white solid in 65.6% yield. 1 H NMR(400MHz,DMSO-d 6 )δ8.91(s,2H),8.09(d,J=1.8Hz,1H),7.61(dd,J=8.3,1.8Hz,1H),7.45(d,J=8.3Hz,1H),3.96(s,3H),2.35=4(td,J=7.1,2.7Hz,4H),2.05(s,1H).
The FTO-4 was prepared by the synthetic method reported in document Broad spectrum anti-cancer compounds for treatment of cancer.
Step 2, under argon atmosphere, FTO-4-alkyne (0.61 g,1.8 mmol) prepared in step 1 and Ru-N are mixed 3 (1.49 g,3 mmol), copper sulfate pentahydrate (0.34 g,1.50 mmol) and sodium ascorbate (0.40 g,2.0 mmol) were dissolved in DMF and stirred for 10h, after the reaction was completed, the solvent was removed by rotary evaporation, and the orange-yellow product FTO-4-Ru was obtained by column chromatography separation in 75.2% yield. 1 H NMR(400MHz,DMSO-d 6 )δ9.50(d,J=5.9Hz,1H),9.08(s,2H),8.91(s,2H),8.63(s,1H),8.52(s,1H),8.23(s,1H),8.09(d,J=1.8Hz,1H),8.02(dd,J=8.0,1.7Hz,1H),7.98–7.90(m,2H),7.89–7.2(m,1H),7.64(d,J=5.9Hz,1H),7.61(dd,J=8.3,1.8Hz,1H),7.45(d,J=8.3Hz,1H),7.42(d,J=8.3Hz,1H),7.31–7.24(m,2H),7.18(d,J=2.1Hz,2H),6.94(s,1H),6.61(d,J=2.4Hz,1H),6.78(ddd,J=8.2,7.1,1.1Hz,1H),6.00–5.93(m,4H),5.34(s,2H),3.96(s,3H),2.78(td,J=7.1,2.7Hz,4H),2.54(s,1H),2.43(d,J=0.7Hz,3H),2.35=4(td,J=7.1,2.7Hz,4H),2.13(s,3H),2.05(s,1H),0.95(t,J=7.0Hz,6H).
Example 6
The preparation method of the metal complex FL1-Ru comprises the following steps:
Figure BDA0004088110750000101
step 1, in argon atmospherePentynoic acid (29.4 mg,0.3 mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (57.5 mg,0.3 mmol) and triethylamine (50. Mu.L) were dissolved in DMF under argon atmosphere, reacted under ice bath for 30min, N-hydroxybenzotriazole (32.4 mg,0.24 mmol) was dissolved in DMF and added dropwise to the above mixture, reacted under ice bath for 15min, 5-aminofluorescein (FL 1) (0.42 g,1.2 mmol) was added to the mixture, the reaction mass was warmed to room temperature, after stirring overnight, the organic solvent was removed, and purified by column chromatography to give FL1-alkyne as a white solid in 69.5%. 1 H NMR(400MHz,Methanol-d 4 )δ7.16(d,J=2.1Hz,2H),7.10(d,J=2.2Hz,1H),7.08(d,J=2.2Hz,1H),6.93(s,1H),6.91(s,1H),6.67(s,1H),6.61(d,J=2.4Hz,1H),6.58(d,J=2.4Hz,1H),2.63(td,J=7.1,2.7Hz,4H),2.05(s,1H).
Step 2, under argon atmosphere, FL1-alkyne (0.77 g,1.8 mmol) obtained in step 1 and Ru-N were subjected to a vacuum treatment 3 (1.49 g,3 mmol), copper sulfate pentahydrate (0.34 g,1.50 mmol) and sodium ascorbate (0.40 g,2.0 mmol) were dissolved in DMF and stirred for 10h, after the reaction was completed, the solvent was removed by rotary evaporation, and the orange yellow product FL1-Ru was obtained by column chromatography separation in 85.2% yield. 1 H NMR(400MHz,DMSO-d 6 )δ9.50(d,J=5.9Hz,1H),9.08(s,2H),8.63(s,1H),8.52(s,1H),8.23(s,1H),8.02(dd,J=8.0,1.7Hz,1H),7.98–7.90(m,2H),7.89–7.2(m,1H),7.64(d,J=5.9Hz,1H),7.42(d,J=8.3Hz,1H),7.31–7.24(m,2H),7.18(d,J=2.1Hz,2H),6.94(s,1H),6.61(d,J=2.4Hz,1H),6.78(ddd,J=8.2,7.1,1.1Hz,1H),6.00–5.93(m,4H),5.34(s,2H),2.78(td,J=7.1,2.7Hz,4H),2.54(s,1H),2.43(d,J=0.7Hz,3H),2.13(s,3H),0.96(t,J=7.0Hz,6H).
Example 7
The preparation method of the metal complex FL2-Ru comprises the following steps:
Figure BDA0004088110750000102
step 1, 2',7' -dichlorofluorescein (FL 2) (0.48 g,1.2 mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride under argon atmosphere(57.5 mg,0.3 mmol) and triethylamine (50. Mu.L) were dissolved in DMF and reacted under ice-bath conditions for 30 minutes, N-hydroxybenzotriazole (32.4 mg,0.24 mmol) was dissolved in DMF and added dropwise to the above mixture, reacted under ice-bath conditions for 15 minutes, propargylamine (82.6 mg,1.5 mmol) was added dropwise to the mixture, the reaction mass was allowed to warm to room temperature overnight, after stirring overnight, the organic solvent was removed, and purified by column chromatography to give FL2-alkyne as a white solid in 77.8% yield. 1 H NMR(400MHz,DMSO-d 6 )δ11.38(s,2H),8.04–8.00(m,1H),7.83(td,J=7.5,1.2Hz,1H),7.76(td,J=7.5,1.0Hz,1H),7.35(d,J=7.7Hz,1H),7.07(s,2H),6.63(s,2H),4.88(s,2H),2.05(s,1H).
Step 2, under argon atmosphere, FL2-alkyne (0.79 g,1.8 mmol) obtained in step 1 and Ru-N were subjected to a vacuum treatment 3 (1.49 g,3 mmol), copper sulfate pentahydrate (0.34 g,1.50 mmol) and sodium ascorbate (0.40 g,2.0 mmol) were dissolved in DMF and stirred for 10h, after the reaction was completed, the solvent was removed by rotary evaporation, and the orange yellow product FL2-Ru was obtained by column chromatography separation in 78.6% yield. 1 H NMR(400MHz,DMSO-d 6 )δ11.38(s,2H),9.50(d,J=5.9Hz,1H),8.63(s,1H),8.09(d,J=1.8Hz,1H),8.04–8.00(m,1H),7.98–7.90(m,2H),7.64(d,J=5.9Hz,1H),7.61(dd,J=8.3,1.8Hz,1H),7.31–7.24(m,2H),7.07(s,2H),6.00–5.93(m,4H),4.88(s,2H),2.78(td,J=7.1,2.7Hz,4H),2.54(s,1H),2.43(d,J=0.7Hz,3H),2.13(s,3H),2.05(s,1H),0.95(t,J=7.0Hz,6H).
Example 8
The preparation method of the metal complex MA-Ru comprises the following steps:
Figure BDA0004088110750000111
step 1, meclofenamic Acid (MA) (0.36 g,1.2 mmol), 1- (3-dimethylaminopropyl) -3-ethylcarbodiimide hydrochloride (57.5 mg,0.3 mmol) and triethylamine (50. Mu.L) were dissolved in DMF under argon atmosphere, reacted under ice bath for 30min, N-hydroxybenzotriazole (32.4 mg,0.24 mmol) was dissolved in DMF and added dropwise to the above mixture, reacted under ice bath for 15min, propargylamine (8) was added dropwise to the mixed solution2.6mg,1.5 mmol), the reaction mass was warmed to room temperature overnight, stirred overnight, the organic solvent removed and purified by column chromatography to give MA-alkyne as a white solid in 72.5% yield. 1 H NMR(400MHz,Methanol-d 4 )δ8.00(dd,J=8.0,1.7Hz,1H),7.41(d,J=8.3Hz,1H),7.31–7.23(m,2H),6.76(ddd,J=8.2,7.1,1.1Hz,1H),6.25(dd,J=8.4,1.0Hz,1H),4.88(s,2H),3.08(s,1H),2.43(d,J=0.7Hz,3H).
Step 2, under argon atmosphere, MA-alkyne (0.60 g,1.8 mmol) prepared in step 1 and Ru-N 3 (1.49 g,3 mmol), copper sulfate pentahydrate (0.34 g,1.50 mmol) and sodium ascorbate (0.40 g,2.0 mmol) were dissolved in DMF and stirred for 10h, after the reaction was completed, the solvent was removed by rotary evaporation, and the orange-yellow product MA-Ru was obtained by column chromatography separation in 80.2% yield. 1 H NMR(400MHz,DMSO-d 6 ))δ9.50(d,J=5.9Hz,1H),9.37(d,J=5.8Hz,1H),9.06(s,2H),8.64(s,1H),8.54(s,1H),8.48(s,1H),8.25(s,1H),8.00(dd,J=8.0,1.7Hz,1H),7.97–7.90(m,2H),7.89–7.83(m,1H),7.66(d,J=5.9Hz,1H),7.41(d,J=8.3Hz,1H),7.31–7.23(m,2H),6.76(ddd,J=8.2,7.1,1.1Hz,1H),6.25(dd,J=8.4,1.0Hz,1H),6.00–5.91(m,4H),5.35(s,2H),4.88(s,2H),3.08(s,1H),2.43(d,J=0.7Hz,3H),2.16(s,3H),0.95(t,J=7.0Hz,6H).
The metal complex of the invention is applied to the aspect of tumor cell treatment.
The method comprises the following steps: MTT colorimetric method and CCK-8 colorimetric method. The present experiment measures the anti-cancer activity in vitro on human cancer cell lines such as human breast cancer cells (MCF-7), mouse-derived brain glioma cells (A261), human chronic myelogenous leukemia cells (K562), human acute promyelocytic leukemia cells (NB-4) and human normal hepatocytes (LO 2). MCF-7, MDA-MB-231 and LO2 cells in DMEM medium containing 10% fetal bovine serum, 1% penicillin-streptomycin solution, K562, NB-4 cells in 1640 medium containing 10% fetal bovine serum, 1% penicillin-streptomycin solution, 5% CO at 37 ℃C 2 Culturing in a cell culture incubator. Inoculating cells into 96-well cell culture plate according to 5000 cells/well initial density, culturing for 24 hr, adding different concentration gradient medicine culture medium, culturing for 48 hr, spreading MCF-7, A261 and LO2 cell well plates, eachmu.L of MTT aqueous solution (5 mg/mL) was added to the wells, incubation was continued for 4h, medium was removed, 150. Mu.L of DMSO was added, shaking was performed until the purple crystals were completely dissolved, and the wells were allowed to shake for 1 minute using a microplate reader (model Tecan Infinite M1000 Pro) and absorbance at 490nm was read. And K562 and NB-4 cell well plates were plated, 10. Mu.L of CCK-8 was added to each well plate, incubation was continued for 3h, and the plates were directly shaken for 1 minute using a microplate reader (model Tecan Infinite M1000 Pro) and absorbance at 450nm was read.
The anticancer activities of the FTO-2 ruthenium/iridium/rhodium complexes prepared in examples 1 to 3 are shown in Table 1.
Table 1 shows the IC's of the complexes FTO-2-Ru, FTO-2-Ir, FTO-2-Rh, FTO-2-alkyne and Cisplatin (CDDP) 50 Value of (. Mu.M)
Figure BDA0004088110750000121
/>
Figure BDA0004088110750000131
The results show that: the organic active molecule FTO-2 and FTO-2-alkyne have little activity on several cells (IC 50 >100. Mu.M), FTO-2-Ru showed low toxicity to several cells but excellent selectivity to NB-4 cells, FTO-2-Ir and FTO-2-Rh showed low toxicity to adherent cells but excellent selectivity to leukemia cells, especially NB-4 cells, while having poor activity to human normal hepatocytes (LO 2), indicating very high selectivity to leukemia cells based on the FTO-2 aryl, metallocene metal complex. Therefore, the FTO-2-Ru, the FTO-2-Ir and the FTO-2-Rh are all beneficial to reducing the side effect of the medicine and have better anticancer activity. The complex of the invention has certain selectivity on leukemia, and can realize targeted treatment on leukemia cells.
Example 9
Positioning of the FTO-2-Ru/Ir/Rh complexes prepared in examples 1-3 of the present invention in cells.
The method comprises the following steps: NB-4 cells were inoculated into a 10cm dish and cultured for 24 hours, and then 1640 culture solution containing FTO-2-Ru (3. Mu.M), FTO-2-Ir (5. Mu.M) and FTO-2-Rh (5. Mu.M) was added thereto, respectively, followed by further culturing for 12 hours. Cells were collected and washed 2 times with PBS (4 ℃). Nuclei, mitochondria and cytoplasm were removed using a mitochondrial/cell nucleus preparation kit, and then each sample was digested with concentrated nitric acid (100. Mu.L, 95 ℃) for 2 hours, hydrogen peroxide (50. Mu.L, 95 ℃) for 1 hour, and concentrated hydrochloric acid (100. Mu.L, 95 ℃) for 2 hours in sequence. After cooling, the resulting liquid was diluted to 2mL with ultrapure water, the metal content in the samples was measured by ICP-MS, and the intracellular metal content was normalized by the number of cells in each sample.
The intracellular localization of the FTO-2-Ru complex of example 1 is shown in FIG. 1A, after the complex FTO-2-Ru (3 mu M) and NB-4 cells are incubated for 12h, organelles are extracted, and the content of metal Ru in samples in different organelles is determined by inductively coupled plasma mass spectrometry ICP-MS, wherein the figure shows that the complex FTO-2-Ru is mainly distributed in the nucleus and mitochondria and partially distributed in cytoplasm. Example 2 intracellular localization of FTO-2-Ir complex as shown in fig. 1B, after 12h incubation of complex FTO-2-Ir (5 μm) with NB-4 cells, organelles were extracted and the content of metallic Ir in samples from different organelles was determined by inductively coupled plasma mass spectrometry ICP-MS, which indicated that the complex FTO-2-Ir was mainly distributed in mitochondria. Example 3 intracellular localization of FTO-2-Rh complex the complex FTO-2-Rh (5 μm) was incubated with NB-4 cells for 12h, the organelles were extracted and the content of metallic Rh in the samples of the different organelles was determined by inductively coupled plasma mass spectrometry ICP-MS, which indicated that the complex FTO-2-Rh was distributed mainly in the nucleus of the cells.
The results show that: the complex FTO-2-Ru/Ir/Rh can be taken up by NB-4 cells in a large amount through cell membranes in a short time, wherein the FTO-2-Ru is mainly distributed in cell nuclei and mitochondria, the FTO-2-Ir is mainly distributed in mitochondria, and the FTO-2-Rh is mainly distributed in cell nuclei.
Example 10
The FTO-2-Ru/Ir/Rh prepared in examples 1-3 of the present invention induced apoptosis.
The method comprises the following steps: NB-4 cells were inoculated into six well plates and grown for 18h, and complexes FTO-2-Ru, FTO-2-Ir and FTO-2-Rh were added at different concentrations. Incubation was carried out for 24h, washed twice with PBS, cells were collected and resuspended in 500. Mu.L Binding buffer, 5. Mu.L Annexin V-FITC was added and mixed well. After 5 minutes, 5 μ L Propidium Iodide was added and mixed well and incubated in the dark for 15 minutes, the samples were analysed by BD FACSverse flow cytometer and the data was analysed by flowjo7.6 software.
The apoptosis induction of the complexes FTO-2-Ru, FTO-2-Ir and FTO-2-Rh of example 1 and example 2 is shown in figure 2, the flow chart of the complexes FTO-2-Ru, FTO-2-Ir and FTO-2-Rh induced NB-4 apoptosis and the apoptosis each phase percentage chart of the cells under different concentration gradients after 24h administration, and the data in the chart show that the complexes FTO-2-Ru cannot induce apoptosis and the complexes FTO-2-Ir and FTO-2-Rh gradually undergo apoptosis along with the increase of the administration concentration.
The results show that: the complex FTO-2-Ru can not induce apoptosis, and the complex FTO-2-Ir and the complex FTO-2-Rh can induce apoptosis to different degrees.
Example 11
The FTO-2-Ru/Ir/Rh prepared in the embodiments 1-3 of the invention promotes the generation of intracellular active oxygen;
the method comprises the following steps: confocal microscopy detects ROS production: NB-4 cells were seeded in confocal dishes and incubated for 12h at 37 ℃. At and 2IC 50 After 24h incubation with FTO-2-Ru, FTO-2-Ir and FTO-2-Rh, the cells were washed twice with PBS. Cells were exposed to fluorescent probe 2',7' -dichlorofluorescein diacetate (DCFH-DA, 10. Mu.M) for 20 min at 37 ℃. The cells were then washed twice with PBS and imaged under a confocal laser microscope.
Flow cytometry detects ROS production: NB-4 cells were seeded in 6-well plates and incubated at 37℃for 12h. At and 2IC 50 After 24h incubation with FTO-2-Ru, FTO-2-Ir and FTO-2-Rh, the cells were washed twice with PBS. Cells were exposed to fluorescent probe 2',7' -dichlorofluorescein diacetate (DCFH-DA, 10. Mu.M) for 20 min at 37 ℃. Cells were collected after washing 2 times with PBS and resuspended with PBS. Immediately after detection using a BD FACSverse flow cytometer, excitation wavelength was 488nm, emission wavelength was 510-540 nm, and data were processed using FlowJo7.6 software.
Implementation of the embodimentsThe complexes FTO-2-Ru of example 1, FTO-2-Ir of example 2 and FTO-2-Rh of example 3 all promote reactive oxygen species formation to varying degrees, as shown in FIG. 3, NB-4 cells were isolated from 2IC 50 After 24h of co-incubation of FTO-2-Ru, FTO-2-Ir and FTO-2-Rh at the concentrations, the complex-induced intracellular reactive oxygen species production was detected by flow cytometry and confocal laser. As can be seen from the data, when the complex is added for 24 hours, active oxygen is produced in the cell.
The results show that: FTO-2-Ru, FTO-2-Ir and FTO-2-Rh can promote the generation of active oxygen to different degrees.
Example 12
FTO-2-Ru, FTO-2-Ir and FTO-2-Rh prepared in examples 1-3 of the invention induce mitochondrial membrane potential decrease;
the method comprises the following steps: NB-4 cells were inoculated into 6-well plates and grown for 12h, and complexes FTO-2-Ru, FTO-2-Ir and FTO-2-Rh with different concentration gradients were added. Incubation for 24h, aspiration of culture medium, pancreatin digestion, harvesting of cells, washing three times with PBS, followed by staining for 30min with formulated JC-1 working solution. The cells were resuspended with JC-1 staining buffer and immediately examined using BD FACSverse flow cytometer, and the two channels were set as FL1 and FL2, respectively, and the data were processed and analyzed using FlowJo7.6 software.
The mitochondrial membrane potential drop induced by the complexes FTO-2-Ru of example 1, FTO-2-Ir of example 2 and FTO-2-Rh of example 3 is shown in FIG. 4, and the mitochondrial membrane potential of NB-4 cells changes after incubation for 24h in different concentrations of the complexes FTO-2-Ru, FTO-2-Ir and FTO-2-Rh. It can be seen that the mitochondrial membrane potential tends to decrease with increasing drug concentration, indicating that the complexes FTO-2-Ru, FTO-2-Ir and FTO-2-Rh can induce the decrease of the mitochondrial membrane potential of NB-4 cells at different degrees.
The results show that: the complexes FTO-2-Ru, FTO-2-Ir and FTO-2-Rh can induce the mitochondrial membrane potential to be reduced to different degrees.
Example 13
The cell cycle retardation phenomena of FTO-2-Ru, FTO-2-Ir and FTO-2-Rh prepared in examples 1-3 of the present invention.
The method comprises the following steps: will beNB-4 cells were seeded in 6-well cell plates at 5% CO 2 After 18 hours of incubation at 37℃the complexes FTO-2-Ru, FTO-2-Ir and FTO-2-Rh were added in different concentration gradients. After 24 hours of treatment at 37 ℃, cells were collected, washed twice with cold PBS and fixed overnight with 70% ethanol at 4 ℃. The fixed mixture was washed 2 times with PBS, pretreated with RNase A (100. Mu.g/mL) for 10min, incubated with propidium iodide (PI, 50. Mu.g/mL) for 30min, washed 2 times with PBS, and then analyzed by flow cytometry to assess the effect of NB-4 on cell cycle arrest.
The complexes FTO-2-Ru of example 1, FTO-2-Ir of example 2 and FTO-2-Rh of example 3 block the cell cycle at G1/G2 as shown in FIG. 5, and after 24h administration, the complexes FTO-2-Ru, FTO-2-Ir and FTO-2-Rh at different concentration gradients have a flow chart for the effect of blocking the NB-4 cell cycle, and it can be seen that the complexes FTO-2-Ru cannot block the cell cycle, and that as the concentrations of the complexes FTO-2-Ir and FTO-2-Rh increase, more cells at G1/G2 phase, and thus the complexes FTO-2-Ir and FTO-2-Rh can block NB-4 cells at G1/G2.
The results show that: the complex FTO-2-Ru cannot block the cell cycle, and FTO-2-Ir and FTO-2-Rh can block the cell cycle in the G1/G2 phase.
Example 14
The application of organic active molecules FTO-2, FTO-2-alkyne, complex FTO-2-Ru, FTO-2-Ir and FTO-2-Rh to inhibit FTO activity.
The method comprises the following steps: 0.1nmol of the mixture contains m 6 ssDNA of A,0.1nmol of FTO, 300. Mu.M of 2OG, 280. Mu.M (NH) 4 ) 2 Fe(SO 4 ) 2 2mM L-ascorbic acid, 100. Mu.M of FTO-2, FTO-2-alkyne, complex FTO-2-Ru, FTO-2-Ir and FTO-2-Rh are dissolved in 100. Mu.L of Tris-HCl (50 mM, PH=7.5) buffer solution, incubated for 2 hours at room temperature, boiled for 5 minutes at 90 ℃ and annealed, 20% of non-reducing PAGE is added, and after electrophoresis run, the gel red staining is used, and the stronger the band strength and the stronger the intensity are compared, the more obvious the inhibition effect is.
FTO-2, FTO-2-alkyne, the FTO-2-Ru of the complex, FTO-2-Ir and FTO-2-Rh have FTO inhibition activity as shown in figure 6, and it can be seen from the figure that FTO-2 and FTO-2-alkyne have no inhibition activity, the complex FTO-2-Ru has the function of degrading DNA, and the FTO-2-Ir and the FTO-2-Rh have FTO inhibition activity.
The results show that: FTO-2-Ir and FTO-2-Rh have FTO inhibitory activity.

Claims (10)

1. A metal complex with FTO inhibitory activity, characterized in that the structural general formula of the complex is shown as follows:
Figure FDA0004088110740000011
wherein M is Ru, ir, rh or Os;
Figure FDA0004088110740000012
is->
Figure FDA0004088110740000013
L is
Figure FDA0004088110740000014
2. The method for producing a metal complex having FTO inhibitory activity as claimed in claim 1, comprising the steps of:
(1) Preparation of azide group-containing metal complex precursors: under inert atmosphere, dissolving a metal ruthenium dimer, a metal iridium dimer, a metal rhodium dimer or a metal osmium dimer in methanol, carrying out coordination reaction with a chelating ligand, removing a solvent by reduced pressure rotary evaporation, adding a methanol solution of saturated anion salt, freezing at low temperature, centrifuging after freezing, collecting solids, and purifying a crude product by column chromatography to obtain a metal complex precursor;
(2) Preparation of alkynyl-modified organic active molecule ligands: under inert atmosphere, placing organic active molecules, alkynyl compounds and catalysts into an organic solvent for reaction, removing the solvent by rotary evaporation after the reaction is finished, and purifying a crude product by column chromatography to obtain an organic active molecule ligand;
(3) And (3) reacting the metal complex precursor, the organic active molecular ligand and the catalyst in an organic solvent under inert atmosphere, removing the solvent by rotary evaporation after the reaction is finished, and purifying the crude product by a chromatographic column to obtain the metal complex.
3. The method for producing a metal complex having FTO inhibitory activity according to claim 2, wherein: in the step (1), the molar ratio of the dimer of the metallic ruthenium to the chelating ligand is 8-9: 1, a step of; the molar ratio of the dimer of the metallic iridium to the chelating ligand is 13-14: 1, a step of; the molar ratio of the dimer of the metal rhodium to the chelating ligand is 12-13: 1, a step of; the molar ratio of the dimer of the metal osmium to the chelating ligand is 12-13: 1.
4. the method for producing a metal complex having FTO inhibitory activity according to claim 2, wherein: in step (1), the chelating ligand is 4-azidomethyl-4 '-methyl-2, 2' -bipyridine.
5. The method for producing a metal complex having FTO inhibitory activity according to claim 2, wherein: in the step (1), the freezing temperature is between-20 and 0 ℃ and the freezing time is between 8 and 14 hours.
6. The method for producing a metal complex having FTO inhibitory activity according to claim 2, wherein: in the step (2), the alkynyl compound is bromopropyne, propargylamine or pentynoic acid; the catalyst is potassium carbonate or N-hydroxybenzotriazole and N, N-diisopropylethylamine according to the mol ratio of 82-83: 135, and a combination thereof.
7. The method for producing a metal complex having FTO inhibitory activity according to claim 2, wherein: in the step (2), the molar ratio of the organic active molecule to the alkynyl compound is 0.1-4: 1.
8. the method for producing a metal complex having FTO inhibitory activity according to claim 2, wherein: in the step (3), the catalyst is cupric sulfate pentahydrate and sodium ascorbate according to the mole ratio of 3-3.5: 4.
9. The method for producing a metal complex having FTO inhibitory activity according to claim 2, wherein: in the step (3), the reaction mole ratio of the metal complex precursor and the organic active molecular ligand is 3-25.5: 1.
10. the use of the metal complex having FTO inhibitory activity as defined in claim 1 for the preparation of an antitumor drug, an antitumor drug component, an FTO inhibitory drug and an FTO inhibitory drug component.
CN202310142825.6A 2023-02-21 2023-02-21 Metal complex with FTO inhibition activity and preparation method and application thereof Pending CN116143843A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310142825.6A CN116143843A (en) 2023-02-21 2023-02-21 Metal complex with FTO inhibition activity and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310142825.6A CN116143843A (en) 2023-02-21 2023-02-21 Metal complex with FTO inhibition activity and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN116143843A true CN116143843A (en) 2023-05-23

Family

ID=86354020

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310142825.6A Pending CN116143843A (en) 2023-02-21 2023-02-21 Metal complex with FTO inhibition activity and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN116143843A (en)

Similar Documents

Publication Publication Date Title
CN113683557B (en) Application of cyclopentadienyl iridium/rhodium dimer
Sarkar et al. Luminescent anticancer ruthenium (II)-p-cymene complexes of extended imidazophenanthroline ligands: synthesis, structure, reactivity, biomolecular interactions and live cell imaging
CN108250250B (en) Complex containing 1,1, 1-triphenyl-N- (1- (pyridine-2-) methylene) methylamine, preparation method and application
Liu et al. Fluorescent iridium (III) coumarin-salicylaldehyde Schiff base compounds as lysosome-targeted antitumor agents
CN114149470B (en) Ruthenium nitrosyl (II) complex and preparation method and application thereof
CN111072726A (en) Synthetic method and application of ruthenium (II) polypyridine metal complex with anti-tumor effect
CN109796503B (en) Preparation method of dinuclear metallocene ruthenium complex and application of dinuclear metallocene ruthenium complex in tumor resistance
CN102942595A (en) Ruthenium complexes, preparation method thereof and application
CN111217861A (en) Iron-iridium heteronuclear metal complex and preparation method and application thereof
CN114524853B (en) All-trans retinoic acid-aryl metal complex, preparation method and application
CN107827934B (en) Tetravalent platinum complex with anticancer activity, preparation method and application
CN116143843A (en) Metal complex with FTO inhibition activity and preparation method and application thereof
Ghasemi et al. Experimental and molecular docking investigation of anticancer activity of new mixed-ligand Schiff base complexes against human colorectal (HCT116), lung (A549) and breast (MCF7) carcinoma cell lines
CN111747889B (en) NNO type quinoline Fe (II) complex containing multiple coordination sites and preparation method and application thereof
CN113072593B (en) Metal organic iridium complex with anticancer activity and preparation method and application thereof
CN111004145B (en) Chiral optical amide substituted alpha, beta-diamino acid derivative and preparation method and application thereof
CN109232670A (en) A kind of preparation method and its antitumor application thereof of cyclopentadienyl type ruthenium complex
CN111718326B (en) Quinoline structure-containing NNN-type pyrimidine hydrazone cobalt (II) complex and preparation method and application thereof
CN113372393B (en) Pt complex containing 5-aminofluorescein ligand and preparation method and application thereof
Liu et al. Synthesis and cancer cell cytotoxicity of gold (III) tetraarylporphyrins with a C5-carboxylate substituent
CN114835706B (en) N≡N ligand and application thereof
CN113512066B (en) Sorafenib-ruthenium complex and preparation method and application thereof
CN113735915B (en) Heteronuclear metal iridium-rhenium complex and preparation method and application thereof
CN111454232B (en) 1,3, 4-thiadiazine compound and application thereof
CN115677788B (en) Metal organic iridium complex with fluorescence characteristic and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination